Skip to main content

Neuromuscular blockers and their reversal: have we finally found the on-off switches?

Abstract

Background

A nondepolarizing neuromuscular blocking agent (NMBA) with a succinylcholine-like quick onset and offset has been the holy grail of the science of neuromuscular blockade. Although this drug is still elusive, the advent of promising new drug combinations like rocuronium–sugammadex and gantacurium–l-cysteine may achieve the same end result. The type of NMBA; the type, timing, and dose of their reversal drugs; the means of monitoring NMB; and the site of monitoring are potentially on the verge of a paradigm shift.

Main text

A comprehensive search using PubMed and Google Scholar and Medline search was made by using keywords gantacurium, l-cysteine, calabadion, and newer neuromuscular blocking agents for peer-reviewed English language manuscripts published before December 2019. Out of the 97 articles screened, 16 were found to be eligible (original articles) and included in this review.

Conclusion

Quantitative, objective neuromuscular monitoring should be included in the minimum monitoring standards. Gantacurium is a new promising nondepolarizing NMBA with desirable succinylcholine-like onset and duration of action without its side effects. A broad-spectrum reversal agent (calabadion) can be used for both depolarizing and nondepolarizing NMB as well as general anesthetics (etomidate and ketamine). A novel drug (WP [6]) can block the side effects of succinylcholine; all are staring at us from the horizon.

Summary

This is a review article about neuromuscular blocking agents (NMBAs) and the broad-spectrum reversal agents.

A non-depolarizing neuromuscular blocking agent (NMBA) with a succinylcholine-like quick onset and offset has for long been the holy grail of the science of neuromuscular blockade. Although this drug is still elusive, the advent of promising new drug combinations like rocuronium–sugammadex and gantacurium–l-cysteine may achieve same end result. The type of NMBA; the type, timing, and dose of their reversal drugs; the means of monitoring NMB; and the site of monitoring are potentially on the verge of a paradigm shift. A broad-spectrum reversal agent (calabadion) for both depolarizing and non-depolarizing NMBAs as well as general anesthetics (etomidate and ketamine) and a novel drug (WP [6]) to block the side effects of succinylcholine are staring at us from the horizon. A comprehensive search using PubMed and Google Scholar and Medline search was made via using keywords gantacurium, l-cysteine, calabadion, and newer neuromuscular blocking agents for peer-reviewed English language manuscripts published before December 2019. Out of the 97 articles screened, 16 were found to be eligible (original articles) and included in this review.

Quantitative, objective neuromuscular monitoring should be included in the minimum monitoring standards. Gantacurium is a new promising nondepolarizing NMBA with desirable succinylcholine-like onset and duration of action minus the side effects. The new broad-spectrum reversible agent calabadion-2 stands out prominently since it does not only reverse any depth of NMB caused by any NMBA, but can also reverse general anaesthetic induction agents and local anaesthetic toxicity. Human clinical trials should be undertaken on a priority basis to explore these exciting realms.

Background

Evolution of neuromuscular blocking agents (NMBAs) commenced with d-tubocurarine (1942) inspired by Amazon-Indian poison arrows (Raghavendra 2002). Notorious for its histamine release, it paved the way for gallamine, the first synthetic NMBA used clinically (1947), but this was highly nephrotoxic. Succinylcholine (1951) was the first leptocurare used clinically but caused fasciculations or momentary muscle excitation preceding muscle relaxation just like decamethonium the only other member of the depolarizing NMBA group (not reversed by anticholinesterase) (Raghavendra 2002). The then existing nondepolarizing NMBAs were replaced by pancuronium the first aminosteroidal NMBA introduced in 1964 and its congener vecuronium (1984). Neither releases histamine, both have a slow onset and unpredictable duration of action in patients with hepatic/renal impairment, but vecuronium is cardio-stable unlike pancuronium which causes tachycardia attributable to vagolytic action. Atracurium and cisatracurium were introduced in 1981 and 1999 respectively, both with and without histamine release, respectively, are belonging to benzylisoquinolium NMBAs. They are eliminated by Hoffmann degradation at physiological temperature and pH. Unfortunately, all these nondepolarizing NMBAs had a slow onset. When Bowman et al. (1988) demonstrated an inverse relationship between NMBA potency and block onset (Bowman et al. 1988), quick/rapid onset (within a minute) NMBAs mivacurium (1992), rocuronium (1994), and rapacuronium (1999) were developed. Gantacurium and its congeners comprise the most modern additions.

Pharmacological reversal of NMBAs begins with the carbamate group, acetylcholinesterase inhibitor “neostigmine” for all practical purposes and since time immemorial (first clinical use 1931; FDA approval 1939), despite drawbacks. Indirect in action, neostigmine cannot reverse profound NMB. It may induce muscle weakness if injected in large doses subsequent to recovery from NMBA (post-operative recurarization) resulting in postoperative respiratory complications (Murphy et al. 2018; Brull and Kopman 2017). Bradycardia, arrhythmias, salivation, flushing, hypotension, and bronchospasm (cholinergic stimulation) may result if not co-administered with anticholinergics (atropine, glycopyrrolate) (Murphy et al. 2018; Brull and Kopman 2017). The only two other clinically available anticholinesterases are pyridostigmine and edrophonium (Colovic et al. 2013).

The ideal NMBA has rapid-onset, quick-offset, non-cumulative, nondepolarizing action, reversible by an antagonist and devoid of clinically relevant adverse effects (Savarese and Kitz 1975; Raghavendra 2002). Rapid onset assumes greatest importance during difficult face-masking with inadequate ventilation/oxygenation, inability to maintain/protect the airway, and anticipation of deteriorating clinical status of the patient besides emergency and obstetric surgery. Finding an NMBA with neuromuscular properties identical to succinylcholine minus its side effect profile is the holy grail of NMB science. Rocuronium has a comparable onset-time but at the cost of a prolonged duration of action (Lien 2013). Reversal is possible with neostigmine only after roughly 30 min of NMBA administration depending upon the train-of-four ratio (TOFr). A specific reversal agent, sugammadex, can reverse profound degrees of rocuronium- and vecuronium-induced NMB (unlike neostigmine) but has its limitations (Haerter and Simons 2015). Moreover, its lipophilic cavity is not roomy enough to envelope benzylisoquinoliums. A broad-spectrum reversal agent, universal for all NMBA, capable of reversing any depth of NMB, is undergoing human clinical trials only now. Supramolecular chemists have developed a brand-new container, calabadion, with a much larger cavity than cyclodextrins (sugammadex) that can envelope and inactivate benzylisoquinoliums as well (Hoffmann et al. 2013).

We may soon witness a paradigm shift from neostigmine to this promising new agent calabadion-2 that can reverse NMB caused by both benzylisoquinolium and aminosteroid NMBA.

This review comprises novel, quick-onset NMBAs and reversal agents facilitating their quick-offset: the on-off switches for neuromuscular block. A comprehensive PubMed, MEDLINE, and Google Scholar search using keywords gantacurium, l-cysteine, calabadion, and newer neuromuscular blocking agents was made for peer-reviewed English language manuscripts published before December 2019, and reference crawling was done. Out of the 97 articles screened, 16 were found to be eligible (original articles) and included in this review.

Despite wide varieties of available NMBAs (Fig. 1), quest for the ideal NMBA is still going on. Quick-onset NMBAs will be briefly discussed focussing on mivacurium, gantacurium, and analogs.

Fig. 1
figure 1

Classification of neuromuscular blocking agents with their respective reversal drugs. (#Obsolete; *reversed with Hoffmann degradation; **reversed with sugammadex)

Main text

Mivacurium (Mivacron; Abbott Laboratories Inc.)

Mivacurium comprises a choline-like bis-benzyl-tetrahydro-isoquinolinium diesteric nondepolarizing NMBA. Spatial orientation of the methylated phenolic moiety results in three (cis-trans, trans-trans, cis-cis) stereoisomers (Lien 2013). It undergoes butyrylcholinesterase metabolism albeit slower than succinylcholine. Although mivacurium is the shortest acting nondepolarizing NMBA available, its duration of action is slightly longer than that of succinylcholine. Despite producing 100% block at laryngeal adductors within 2.5 min and the recovery index being 6 min versus 15 min for atracurium and 30 min for vecuronium infusions, mivacurium is still not popular (Diefenbach et al. 1995). No tachyphylaxis or phase-2 block is seen after prolonged infusion. A major drawback of mivacurium is possible inadequate intubating conditions after a 2 × ED95 dose since mivacurium metabolism begins while a block is still developing. FDA approval was obtained in 1992, but Abbott ceased marketing mivacurium in the USA in 2006 due to loss of a chemical intermediary supplier. Since this was not due to safety or efficacy concerns, FDA has placed mivacron (2 mg equivalent/ml) under “discontinued drug product list” section of the orange book (United States Food and Drug Administration n.d.).

Gantacurium (AV430A; GW280430A)

This olefinic compound signifies the birth of a new generation of NMBAs. Chemically, gantacurium is an asymmetric, enantiomeric, isoquinoliniumchlorofumaric acid diester. Gantacurium is a single isomer just like cisatracurium (unlike mixed-isomers atracurium and mivacurium) (Savarese et al. 2010; Boer and Carlos 2018; Heerdt et al. 2015; Lien et al. 2009) and needs reconstitution before administration (Heerdt et al. 2015).

Gantacurium (Table 1) is a rapid onset, nondepolarizing NMBA currently undergoing clinical trials. Intravenous l-cysteine can reverse gantacurium blockade of any depth akin to sugammadex reversal of rocuronium. Compared with the depolarizing muscle relaxant succinylcholine, gantacurium (2–3 × ED95) causes a 100% neuromuscular block at the laryngeal adductors within 60 s, whereas succinylcholine (3 × ED95) reaches its maximal effect in 45 s (Boer and Carlos 2018). Spontaneous recovery after gantacurium (2 × ED95)-induced neuromuscular block mimics that of succinylcholine-induced neuromuscular block without unwanted succinylcholine side effects. However, gantacurium is not yet available in clinical practice.

Table 1 Analysis of in vivo studies involving novel, quick-onset NMBAs and reversal agents facilitating their quick-offset: the on-off switches

Akin to cisatracurium and atracurium inactivation by Hofmann elimination, gantacurium is metabolized by spontaneous cysteine adduction (fast process) and pH-sensitive hydrolysis (slow process). Former is independent of the liver, kidneys, pH, or temperature. Endogenous L-cysteine replaces the chlorine moiety of gantacurium producing a heterocyclic ring which does not interact with the post-junctional acetylcholine receptors (Lien et al. 2009).

Exogenous l-cystine enantiomer is an essential constituent of parental nutrition. A bolus dose of 10–50 mg/kg for reversal of gantacurium-induced NMB has no known toxicity. In preclinical trials, l-cysteine administered just 1 min after 8 × ED95 of gantacurium reduced the recovery to a TOFr ≥ 0.90 by 6 min without any signs of residual NMB or recurarization. These results need clinical validation. Metabolites of gantacurium lack neuromuscular properties with no hepatorenal elimination (Savarese et al. 2010; Boer and Carlos 2018; Heerdt et al. 2015; Lien et al. 2009).

Cholinesterase inhibitor edrophonium has a peak effect of less than 2 min (Savarese et al. 2010). In humans, edrophonium reduces the reversal time of a gantacurium-induced NMB at 10% recovery of twitch 1 to a TOFr ≥ 0.90 to 3. 8 min. It took 7.5 min for spontaneous reversal of the same NMB. Peak effect being 7–11 min, neostigmine is not suitable for reversal of a gantacurium-induced neuromuscular block (Savarese et al. 2010).

Tables 2 and 3 provides a comparative analysis of early onset NMBAs at different doses expressed as multiples of ED95 (effective dose of NMBA required to reduce twitch height by 95%). Intubating dose is roughly twice the ED95. Block onset time is inversely proportional to NMBA potency. Histamine release by chemical (anaphylactoid) or immunologic (anaphylactic) mechanisms is clinically indistinguishable. NMBAs are the most frequently implicated class of drugs with succinylcholine being the commonest culprit in intraoperative anaphylaxis (Ezzat et al. 2011; Naguib et al. 1995; Spoerl et al. 2017).

Table 2 Comparative analysis of early onset NMBAs at different doses expressed as multiples of ED95 (effective dose 95)
Table 3 Classification, dose, and side effects of reversal agents

CW002

CW002 (Table 1) is a quick-onset intermediate-acting, tetrahydroisoquinolinium nondepolarizing NMBA with minimal cardiopulmonary effects currently undergoing clinical trials (Savarese et al. 2010; Boer and Carlos 2018; Heerdt et al. 2016). It differs from gantacurium in lacking a chlorine-moiety at the fumarate double bond. CW002 undergoes pH-dependent l-cysteine adduction and ester hydrolysis and can be reversed at any depth by exogenous cysteine injection (Boer and Carlos 2018; Heerdt et al. 2016). To date, no human study on exogenous l-cysteine reversal of CW002 exists, but in Rhesus monkeys l-cysteine 50 mg/kg resulted in a reversal of neuromuscular block within 2–3 min when administered 60 s after 4–5× ED95

CW011

This is a non-halogenated olefinic diester congener of gantacurium with a similar onset but intermediate duration of action (Savarese et al. 2010; Boer and Carlos 2018). l-cysteine dose required is 50 mg/kg for antagonism of CW011 as against 10 mg/kg for gantacurium antagonism since chlorine (halogen) substitution in gantacurium is a powerful accelerator of l-cysteine adduction (short t1/2 of 0.2 min) as against t1/2 of 11.4 and 13.7 min in CW002 and CW011 respectively. This explains both the ultrashort duration of onset and offset of gantacurium (Savarese et al. 2010; Boer and Carlos 2018).

CW1759-50

This rapid-onset, ultrashort-acting NMBA claims a superior clinical profile to gantacurium and entails reduced hemodynamic perturbations (Table 1). The half-time of adduction of l-cysteine to CW 1759-50 in vitro is 2.3 min. The ED95 of CW 1759-50 is 0.069 mg/kg which is similar to that of gantacurium (0.081 mg/kg) (Savarese et al. 2018). Human clinical trials on this promising agent are recommended.

Pinnatoxins and 20-methylspirolide-G (20-meSPX-G)

Derived from marine planktons and dinoflagellates, pinnatoxins (macrocyclic imines) and 20-meSPX-G (cyclic amine) are nAchr competitive antagonists, targeting embryonic (α1)2βγδ and adult (α1)2βεδ skeletal muscle neuromuscular junction receptors (Delcourt et al. 2019; Couesnon et al. 2016). 20-meSPX-G is 75 times more potent than d-tubocurarine (Couesnon et al. 2016). In mouse bioassays, the action of 20-meSPX-G is fully reversible producing muscle paralysis but no lethality (Couesnon et al. 2016). A new class of nondepolarizing NMBAs may emerge from here.

Structure activity relationships

The active site (anionic binding region) of postsynaptic nicotinic acetylcholine receptors (nAchR) is similar to that of acetylcholinesterase, and both require a quaternary amine to bind with it. A succinylcholine molecule comprises two acetylcholine molecules linked together because two anionic binding sites of each nAchR need to be simultaneously occupied by two acetylcholine molecules for channel opening (Fig. 2) Succinylcholine and many nondepolarizing NMBA (pancuronium, atracurium) are bis-quaternary ammonium compounds with two quaternary ammonium nitrogen (one for each anionic binding site of nAchR) bridged by 10–12 carbon atoms (for maximal potency). Monoquarternary aminosteroid NMBAs are less potent but have faster onset. Neostigmine is also a quaternary ammonium compound, and hence, it binds acetylcholinesterase. Acetylcholine is hydrolyzed within 100 μs whereas neostigmine acts as a competitive inhibitor of acetylcholine and is hydrolyzed in minutes (40 × 106 times slower). Sugammadex and calabadion are encapsulating reversal agents (Fig. 3).

Fig. 2
figure 2

Structure-activity relationships of acetylcholine, succinylcholine, and anticholinesterase reversal agents

Fig. 3
figure 3

Chemical structure of supramolecular containers sugammadex and calabadion

Five important questions need evidence-based answers before we discuss newer reversal agents.

  1. 1.

    Should a peripheral nerve stimulator (PNS) be used as guide in all patients given NMBA?

  2. 2.

    Should we reverse of NMBA at the end of surgery?

  3. 3.

    What is the optimal timing of reversal agent?

  4. 4.

    What is the optimal dose of reversal agent?

  5. 5.

    Which kind of reversal agent should we use?

There is a convincing evidence that if the anesthesiologists do not reverse NMBA with a reversal agent, it will be translated into a high incidence of post-operative residual NMB. In one study on 568 consecutive patients, 42% of patients in whom vecuronium-induced NMB was not reversed with an anticholinesterase displayed TOFr< 0.7 on postoperative ward arrival (Baillard et al. 2000). Similarly, 57% of patients who received cisatracurium (2 × ED95) and 44% of those receiving rocuronium (2 × ED95) and were not reversed had TOFr < 0.9 on reaching SICU (Maybauer et al. 2007).

Alarmingly, 89% of elderly patients displayed postoperative residual NMB after intraoperative rocuronium administration in one study (Pietraszewski and Gaszyński 2013). Even reversal with sugammadex, if lacking peripheral nerve stimulation (PNS) guidance, does not guarantee protection from residual NMB (Kotake et al. 2013). Even 2 h after a single bolus dose of intermediate-acting NMBA (vecuronium, rocuronium, atracurium), 45% out of 526 consecutive patients showed TOFr< 0.9 when a reversal agent was avoided (Debaene et al. 2003). NMBA residual weakness of the jaw and tongue may cause retention of secretions, aspiration, and pneumonia (Grayling and Sweeney 2007).

Five- second head raise, tongue-protrusion, eye-opening, coughing, and adequacy of tidal volume are frequently used qualitative predictors of recovery from NMBA but cannot exclude clinically significant residual curarization (TOFr 0.5–0.9) (Hemmerling and Le 2007; Hunter 2017). Although most specific, a sustained tongue depressor test has poor sensitivity (18%) for predicting TOFr< 0.9 (Rodney et al. 2015). Measuring grip strength of the dominant hand using electronic hand dynamometer gives a strong correlation (0.89) with TOFr without being distressing to the awake patient (Pei et al. 2019).

Hence, services of a neuromuscular monitor (peripheral nerve stimulator provides only a qualitative assessment) are imperative for quantitative assessment of depth of NMB towards the end of surgery (Brull and Kopman 2017; Gelb et al. 2018). Electromyography is the gold standard, followed by mechanomyography. Nevertheless, acceleromyography and kinemyography (both piezoelectric crystal based) command better clinical utility (Brull and Kopman 2017). Orbicularis oculi and corrugator supercilii, being more centrally located than adductor pollicis, are now being recommended as the preferred site of monitoring NMB onset, for their more faithful representation of NMB onset in the airway musculature (Lien 2013).

Now, tackling the second question, if TOFr is 0.9, then a reversal agent is not required at all. Moreover, if neostigmine or other anticholinesterases are administered at this juncture, it is proved to be not just useless but also counter-productive. Neostigmine may potentially cause reduced genioglossus muscle activity causing increased upper-airway collapsibility in response to negative pharyngeal pressure, recurarization, and upper-airway muscle weakness if given at TOFr 0.9–1, probably by depolarizing or open-channel neuromuscular block and Ach-receptor desensitization (Herbstreit et al. 2010; Eikermann et al. 2008).

Neostigmine dose exceeding 0.07 mg/kg has a ceiling effect. Moreover, excessive neostigmine may precipitate cholinergic crisis with attendant muscle weakness. A decade back, it was realized that neostigmine should be administered after appearance of at least two TOF twitches to reduce postoperative residual paralysis (Brull and Murphy 2010). It is showed that if neostigmine is given at a TOFr of 0.4, then it will assure TOFr 0.9 within 10 min, but not if it is administered earlier (Song et al. 2015). Hence, ideally, neostigmine should be administered only in the window period of TOFr 0.1–0.8, neither before appearance of all four TOF twitches, nor after TOFr 0.9 is achieved. Without PNS availability, making such a fine distinction is difficult. Moreover, time-pressure, anesthesia time, and monetary and human resource factors may prohibit keeping the OT table occupied indefinitely after end of surgery waiting for a spontaneous reversal of NMB. Also, a deeper plane of NMB is required for endoscopic surgery, foreign body removal, and minimally invasive surgery (laparoscopic/robot assisted) although the keyhole port incisions ensure a speedy closure at the end of surgery giving very little time for spontaneous reversal of NMB. A deeper block improves the surgeon satisfaction score (Blobner et al. 2015) by allowing better anatomical exposure at reduced insufflation pressures and avoiding catastrophic patient movement with robotic arms docked. Here comes the role of reversal agents that are direct-acting supramolecular containers: sugammadex and calabadions that can quickly reverse any depth of NMB. The trident of speed, reliability, and safety summarizes the goal for reversal agents.

Sugammadex (gamma cyclodextrin; Bridion; Merck)

Unlike neostigmine, which indirectly reverses NMBA block by increasing acetylcholine concentration at the neuromuscular junction, sugammadex directly inactivates steroidal nondepolarizing NMBAs by effective encapsulation. Sugammadex (2 mg/kg) provides faster reversal (2.7 min versus 17.9 min) of vecuronium-induced neuromuscular blockade compared with neostigmine (50 μg/kg) (Khuenl-Brady et al. 2010). Sugammadex is equally effective in reversing rocuronium-induced block regardless of propofol or sevoflurane anesthesia (Vanacker et al. 2007).

Time to recovery (TOFr0.9) after 2 mg/kg sugammadex administered on appearance of second twitch is 1.9 min and 2.9 min for rocuronium and vecuronium respectively. Similarly, time to recovery after 16 mg/kg sugammadex administered 3 min after 1.2 mg/kg rocuronium is just 1.7 mins (Herring et al. 2017).

Time to spontaneous recovery of first twitch after a single bolus dose of rocuronium was 17 min versus 24 min after rocuronium infusion in most patients. Some patients took 70 min after discontinuing infusion for spontaneous recovery to a TOFr of 75% highlighting the importance of a reversal agent in all cases (Jellish et al. 2000). The additional cost of using sugammadex was estimated at $77/person when compared to neostigmine/glycopyrrolate combination in one study (Money et al. 2019).

Sugammadex 1.0 mg/kg, but not 0.5 mg/kg, adequately reversed a vecuronium-induced NMB at threshold TOF-count of four but without preventing recurarization (Asztalos et al. 2017). Under-dosing of sugammadex as a potential cost-saving strategy in reversal of deep NMB is not recommended as transient success can transcend into disaster like post-operative residual curarization with attendant respiratory complications. Although in use for a decade in Europe and Japan, sugammadex was rejected thrice by the US-FDA on grounds of allergic (Miyazaki et al. 2018; Menéndez-Ozcoidi et al. 2011) and hemorrhagic complications before being accepted in December 2015, despite a relatively high anaphylaxis rate of 1/2580 patients (0.39%) (Miyazaki et al. 2018). Sugammadex prolongs activated partial thromboplastin time and prothrombin time and may cause oral-contraceptive failure (Rahe-Meyer et al. 2014). Potential litigation over side effects is a concern. Although chronic dexamethasone administration induces resistance to NMBAs by augmenting surface and junctional nAchR density, it does not augment sugammadex reversal of rocuronium (Oh et al. 2019).

l-Cysteine

Gantacurium undergoes chemical degradation involving nonessential amino acid, “cysteine” adduction to its central fumarate double bond. In this Michael-type addition reaction, cysteine replaces chlorine to form a heterocyclic ring between the two quaternary heads of gantacurium forming a cysteine adduct with minimal neuromuscular blocking effect (Lien et al. 2009).

Exogenously administered l-cysteine (Sigma-Aldrich, St. Louis, MO; 98% purity; 10–50 mg/kg) (Savarese et al. 2010) can reverse any depth of gantacurium, CW002 and CW011 blockade. l-Cysteine adduction half-life calculated at gantacurium (200 g/ml), CW002 (100 g/ml), and CW011 (50 g/ml) (4:2:1 relative potency ratio) was 0.2 min, 11.4 min, and 13.7 mins respectively, in rhesus monkeys (Heerdt et al. 2015). Cysteine-adduct hydrolysis time was estimated to be 300 min for gantacurium-cysteine adduct and 60 min each for CW002and CW011 respectively.

Calabadions (cucurbit[n]urils n = 5, 6, 7, 8, 10)

Professor Lyle Isaacs established Calabash Biosciences after developing a novel group of molecular containers called calabadions to satisfy the market demand of US anesthesiologists.

Calabadion-1

This is an acyclic, glycoluril, tetrameric, cucurbituril container. Hoffman et al. (2013) administered Calabadion-1 at 30, 60, and 90 mg/kg (for rocuronium; 3.5 mg/kg) and 90, 120, and 150 mg/kg (for cisatracurium 0.6 mg/kg), or neostigmine/glycopyrrolate (0.06/0.012 mg/kg) in rats. The recovery time to TOFr 0.9 was 16.2 min for placebo, 4.6 min for neostigmine, and 84 s for calabadion-1. Cardiopulmonary parameters and blood pH were unaltered. Ninety percent of calabadion-1 was renally excreted within an hour. Calabadion-1-rocuronium complex (Ka = 8.4 ± 0.9 × 106/m) has a comparable binding-constant (affinity) with that of sugammadex-rocuronium complex (Ka = 1.1 ± 0.2 × 107/m), but the binding-constant for calabadion-1-cisatracurium complex is 10-times lesser. Calabadion-1-acetylcholine complex has a binding-constant 350 times smaller than that for calabadion-1-rocuronium. Akin to its predecessor cucurbituril (Haerter and Simons 2015), calabadion-1, forms stable host–guest complexes with local anesthetics in vitro (Ma et al. 2012b).

Calabadion-2 (Calabash Bioscience, Inc. College Park, Maryland)

Haerter et al. (2015) demonstrated through in vitro studies that calabadion-2 (Ka = 3.4 × 109 M− 1) binds rocuronium with 89 times the affinity of sugammadex (Ka = 3.8 × 107 M− 1). The results of proton nuclear magnetic resonance urinalysis, competition binding assays, and ex vivo study (n = 34; phrenic nerve hemidiaphragm preparation) obtained in the absence of metabolic deactivation displayed a 1:1 binding ratio of sugammadex and calabadion-2 toward rocuronium. In live rat models (n = 108; quadriceps femoris muscle), calabadion-2 rapidly reversed 2 × ED90 vecuronium-, rocuronium-, and cisatracurium-induced neuromuscular block in a dose-dependent manner much faster than sugammadex. Calabadion-2 exhibited a higher molar potency to reverse vecuronium and rocuronium, versus sugammadex. Calabadion-2 was eliminated via kidneys, was well tolerated, and had no hemodynamic perturbations. One-hour post-intravenous calabadion-2 (40–80 mg/kg), 49% of the drug was detectable in urine while at lower dosage (5–10 mg/kg), 62% of calabadion-2 appeared.

The enhanced target-binding affinity of calabadion-2 is attributable to its larger hydrophobic cavity shaped by two naphthalene walls versus two benzene walls of calabadion-1 (Zhang et al. 2014). Selectivity of calabadion-2 for rocuronium is 18,900 times that of acetylcholine while that of calabadion-1 is just 350 times that of acetylcholine (same as that of sugammadex).

Ganapati et al. (2016) studied the effect of 27 common drugs (Table 1) on the calabadion-2-NMBA (cisatracurium/rocuronium/vecuronium) complex. Neither the binding affinity nor the standard dosages of these drugs were high enough to displace NMBA from its calabadion-2 container.

Additional benefits of calabadion: a new concept of reversal of general anesthesia

Reversal of general anesthetic induction and maintenance agents and not just NMBAs (Fig. 4) is possible with calabadion-2 potentially translating into time and monetary benefits by slashing operation theater time, reducing postoperative complications, and reversing toxic overdose in hospital and recreational settings.

Fig. 4
figure 4

Classification of neuromuscular block reversal agents

Experiments on rats (Diaz-Gil et al. 2016) demonstrated that calabadion-2 reverses etomidate and ketamine anesthesia by chemical encapsulation at non-toxic plasma concentrations. Electroencephalographic predictors of depth of anesthesia, drug-induced hypotension, recovery of righting-reflex, and functional mobility were studied. Calabadion-2 neither inhibited the human ether-à-go-go-related channel nor was it mutagenic (Ames test). Based on maximum tolerable dose and acceleration of righting reflex recovery, the therapeutic index of calabadion-2 was 16:1 and 3:1 for ketamine and etomidate reversal respectively.

Calabadions seem potentially useful in additional domains like local anesthetic (including cocaine) toxicity (Grabitz et al. 2015; Isaacs et al. 2018).

Water-soluble carboxylatopillar [6] arene (WP [6])

Zhang et al. (2019) studied the antidotal properties of a supramolecular synthetic receptor WP [6] for succinylcholine-induced hyperkalemia, cardiac arrhythmias, rhabdomyolysis, and paralysis in succinylcholine-overdosed mouse models. They reported a reduced incidence of cardiac arrhythmias, hyperkalemia, and muscular damage when WP [6] was injected immediately after succinylcholine explained by reversal of succinylcholine-induced depolarization and diminished efflux of intracellular potassium. It remains to be seen whether and after how much time WP [6] can reverse succinylcholine-induced paralysis if injected simultaneously with succinylcholine in humans.

Conclusion

Quantitative, objective neuromuscular monitoring should be included in the minimum monitoring standards. Gantacurium is a new promising nondepolarizing NMBA with desirable succinylcholine-like onset and duration of action without its side effects. The broad-spectrum reversal agent calabadion-2 stands out prominently since it does not only reverse any depth of NMB caused by any NMBA, but can also reverse general anesthetic induction agents and local anesthetic toxicity. Human clinical trials should be undertaken on a priority basis to explore these exciting realms.

Availability of data and materials

All data related to this review article are contained within the manuscript.

References

  • Asztalos L, Szabó-Maák Z, Gajdos A, Nemes R, Pongrácz A, Lengyel S (2017) Reversal of vecuronium-induced neuromuscular blockade with low-dose sugammadex at train-of-four count of four. A randomized controlled trial. Anesthesiology 127:441–449

    Article  CAS  PubMed  Google Scholar 

  • Baillard C, Gehan G, Reboul-Marty J, Larmignat P, Samama CM, Cupa M (2000) Residual curarization in the recovery room after vecuronium. Br J Anaesth. 84:394–395

    Article  CAS  PubMed  Google Scholar 

  • Belmont MR, Lien CA, Tjan J, Bradley E, Stein B, Patel SS (2004) Clinical pharmacology of GW280430A in humans. Anesthesiology. 100:768–773

    Article  CAS  PubMed  Google Scholar 

  • Blobner M, Frick CGR, Stäuble B (2015) Neuromuscular blockade improves surgical conditions (NISCO). Surg Endosc 26:627–636

    Article  Google Scholar 

  • Blobner M, Mirakhur RK, Wierda JMKH, Wright PMC, Olkkola KT, Debaene B (2000) Rapacuronium 2.0 or 2.5 mg kg–1 for rapid-sequence induction: comparison with succinylcholine 1.0 mg kg–1. Br J Anaesth 85:724–731

    Article  CAS  PubMed  Google Scholar 

  • Boer HD, Carlos RV (2018) New drug developments for neuromuscular blockade and reversal: gantacurium, CW002, CW011, and calabadion. Curr Anesthesiol Rep 8:119–124

    Article  PubMed  PubMed Central  Google Scholar 

  • Bowman WC, Rodger IW, Houston J, Marshall RJ, McIndewar I (1988) Structure: action relationships among some desacetoxy analogues of pancuronium and vecuronium in the anaesthetised cat. Anesthesiology. 69:57–62

    Article  CAS  PubMed  Google Scholar 

  • Brull SJ, Kopman AF (2017) Current status of neuromuscular reversal and monitoring: challenges and opportunities. Anaesthesiology 126:173–190

    Article  Google Scholar 

  • Brull SJ, Murphy GS (2010) Residual neuromuscular block: lessons unlearned. Part II: methods to reduce the risk of residual weakness. Anesth Analg 111:129–140

    Article  PubMed  Google Scholar 

  • Chavan SG, Gangadharan S, Gopakumar AK (2016) Comparison of rocuronium at two different doses and succinylcholine for endotracheal intubation in adult patients for elective surgeries. Saudi J Anaesth 10:379–383

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Colovic MB, Krstic DZ, Lazarevic-Pasti TD, Bondzic AM, Vasic VM (2013) Acetylcholinesterase inhibitors: pharmacology and toxicology. Curr Neuropharmacol 11:315–335

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Couesnon A, Aráoz R, Iorga B, Benoit E, Reynaud M, Servent D (2016) The dinoflagellate toxin 20-methyl spirolide-G potently blocks skeletal muscle and neuronal nicotinic acetylcholine receptors. Toxins. 8:2491–2414

    Article  CAS  Google Scholar 

  • De Mey JC, Debrock M, Rolly G (1994) Evaluation of the onset and intubation conditions of rocuronium bromide. Eur J Anaesthesiol 9:37–40

    Google Scholar 

  • Debaene B, Plaud B, Dilly MP, Donat F (2003) Residual paralysis in the PACU after a single intubating dose of nondepolarizing muscle relaxant with an intermediate duration of action. Anesthesiology. 98:1042–1048

    Article  CAS  PubMed  Google Scholar 

  • Delcourt N, Lagrange E, Abadie E, Fessard V, Frémy JM, Vernoux JP (2019) Pinnatoxins’ deleterious effects on cholinergic networks: from experimental models to human health. Mar Drugs 17:4251–4213

    Article  CAS  Google Scholar 

  • Diaz-Gil D, Haerter F, Falcinelli S, Ganapati S (2016) A novel strategy to reverse general anesthesia by scavenging with the acyclic cucurbit[n]uril-type molecular container calabadion 2. Anesthesiology. 125:333–345

    Article  CAS  PubMed  Google Scholar 

  • Diefenbach C, Buzello W, Mellinghoff H (1995) Mivacurium chloride—a comparative profile. Acta Anaesthesiol Scand. 39:23–25

    Article  Google Scholar 

  • Eikermann M, Zaremba S, Malhotra A, Jordan AS, Rosow C, Chamberlin NL (2008) Neostigmine but not sugammadex impairs upper airway dilator muscle activity and breathing. Br J Anaesth. 101:344–349

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Ezzat A, Fathi E, Zarour A, Singh R, Abusaeda MO, Hussien MM (2011) The optimal succinylcholine dose for intubating emergency patients: retrospective comparative study. Libyan J Med. 6:10–13

    Article  PubMed Central  Google Scholar 

  • Ganapati S, Zavalij PY, Eikermann M, Isaacs L (2016) In vitro selectivity of an acyclic cucurbit[n]uril molecular container towards neuromuscular blocking agents relative to commonly used drugs. Org Biomol Chem 14:1277–1287

    Article  CAS  PubMed  Google Scholar 

  • Gelb AW, Morriss WW, Johnson W, Merry AF, Abayadeera A, Belîi N (2018) World Health Organization-World Federation of Societies of Anaesthesiologists (WHO-WFSA) international standards for a safe practice of anesthesia. Anesth Analg. 126:2047–2055

    Article  PubMed  Google Scholar 

  • Grabitz SD, Ganapati S, Cotten JF, Isaacs L, Eikermann M (2015) Calabadion binds cocaine in vitro and reverses cocaine’s behavioral effects dose-dependently in vivo (abstract A1193). American Society of Anesthesiologists Annual Meeting, Orlando, Florida

    Google Scholar 

  • Grayling M, Sweeney BP (2007) Recovery from neuromuscular blockade: a survey of practice. Anaesthesia. 62:806–809

    Article  CAS  PubMed  Google Scholar 

  • Haerter F, Simons JCP (2015) Comparative effectiveness of calabadion and sugammadex to reverse non-depolarizing neuromuscular-blocking agents. Anesthesiology. 123:1337–1349

    Article  CAS  PubMed  Google Scholar 

  • Heerdt PM, Kang R, Hashim M, Mook RJ, Savarese JJ (2004) Cardiopulmonary effects of the novel neuromuscular blocking drug GW280430A (AV430A) in dogs. Anesthesiology 100:846–851

    Article  CAS  PubMed  Google Scholar 

  • Heerdt PM, Sunaga H, Owen JS, Murrell MT, Malhotra JK, Godfrey D, Steinkamp M, Savard P, Savarese JJ, Lien CA (2016) Dose–response and cardiopulmonary side effects of the novel neuromuscular-blocking drug CW002 in man. Anesthesiology 125:1136–1143

    Article  CAS  PubMed  Google Scholar 

  • Heerdt PM, Sunaga H, Savarese JJ (2015) Novel neuromuscular blocking drugs and antagonists. Curr Opin Anaesthesiol. 28:403–410

    Article  CAS  PubMed  Google Scholar 

  • Hemmerling TM, Le N (2007) Brief review: neuromuscular monitoring: an update for the clinician. Can J Anaesth. 54:58–72

    Article  PubMed  Google Scholar 

  • Herbstreit F, Zigrahn D, Ochterbeck C, Peters J, Eikermann M (2010) Neostigmine/glycopyrrolate administered after recovery from neuromuscular block increases upper airway collapsibility by decreasing genioglossus muscle activity in response to negative pharyngeal pressure. Anesthesiology. 113:1280–1288

    Article  CAS  PubMed  Google Scholar 

  • Herring WJ, Woo T, Assaid CA, Lupinacci RJ, Lemmens HJ, Blobner M (2017) Sugammadex efficacy for reversal of rocuronium- and vecuronium-induced neuromuscular blockade: a pooled analysis of 26 studies. J Clin Anesth. 41:84–91

    Article  CAS  PubMed  Google Scholar 

  • Hoffmann U, Grosse-Sundrup M, Eikermann-Haerter K, Zaremba S, Ayata C, Zhang B, Ma D, Isaacs L, Eikermann M (2013) Calabadion: a new agent to reverse the effects of benzylisoquinoline and steroidal neuromuscular-blocking agents. Anesthesiology. 119:317–325

    Article  CAS  PubMed  Google Scholar 

  • Hunter JM (2017) Reversal of residual neuromuscular block: complications associated with perioperative management of muscle relaxation. Br J Anaesth. 119:53–62

    Article  CAS  Google Scholar 

  • Isaacs LD, Eikermann M, Cusin C, Cotten J, inventors; General Hospital Corp, University of Maryland, College Park, assignee. Acyclic cucurbit[n]uril type molecular containers to treat intoxication and decrease relapse rate in substance abuse disorders. United States patent US 9,956,229. 2018.

    Google Scholar 

  • Jellish WS, Brody M, Sawicki K, Slogoff S (2000) Recovery from neuromuscular blockade after either bolus and prolonged infusions of cisatracurium, or rocuronium using either isoflurane or propofol-based anesthetics. Anesth Analg. 91:1250–1255

    CAS  PubMed  Google Scholar 

  • Kaullen JD, Owen JS, Brouwer KL, Heerdt PM, Lien CA, Savarese JJ, Schmith VD (2018) Pharmacokinetic/pharmacodynamic model of CW002, an investigational intermediate neuromuscular blocking agent, in healthy volunteers. Anesthesiology. 128:1107–1116

    Article  CAS  PubMed  Google Scholar 

  • Khuenl-Brady KS, Wattwil M, Vanacker BF (2010) Sugammadex provides faster reversal of vecuronium-induced neuromuscular blockade compared with neostigmine: a multicenter, randomized, controlled trial. Anesth Analg 110:64–73

    Article  PubMed  Google Scholar 

  • Kotake Y, Ochiai R, Suzuki T, Ogawa S, Takagi S, Ozaki M (2013) Reversal with sugammadex in the absence of monitoring did not preclude residual neuromuscular block. Anesth Analg 117:345–351

    Article  PubMed  Google Scholar 

  • Lien CA (2013) MEikermann, Neuromuscular blockers and reversal agents. In: Hemmings HC, Egan TD (eds) Pharmacology and physiology for anesthesia foundations and clinical practice. Elsevier Saunders, Philadelphia, pp 325–348

    Chapter  Google Scholar 

  • Lien CA, Savard P, Belmont M, Sunaga H, Savarese JJ (2009) Fumarates: unique nondepolarizing neuromuscular blocking agents that are antagonized by cysteine. J Crit Care 24:50–57

    Article  CAS  PubMed  Google Scholar 

  • Ma D, Glassenberg R, Ghosh S, Zavalij PY, Isaacs L (2012b) Acyclic cucurbituril congener binds to local anaesthetics. Supramol Chem. 24:325–332

    Article  CAS  Google Scholar 

  • Ma D, Zhang B, Hoffmann U, Sundrup MG, Eikermann M, Isaacs ML (2012a) Acyclic cucurbit [n] uril-type molecular containers bind neuromuscular blocking agents in vitro and reverse neuromuscular block in vivo. Angew Chem Int Ed 51:11358–11362

    Article  CAS  Google Scholar 

  • Maybauer DM, Geldner G, Blobner MA, Pühringer F, Hofmockel R, Rex C (2007) Incidence and duration of residual paralysis at the end of surgery after multiple administrations of cisatracurium and rocuronium. Anaesthesia 62:12–17

    Article  CAS  PubMed  Google Scholar 

  • Menéndez-Ozcoidi L, Ortiz-Gómez JR, Olaguibel-Ribero JM, Salvador-Bravo MJ (2011) Allergy to low dose sugammadex. Anaesthesia. 66:217–219

    Article  PubMed  Google Scholar 

  • Miyazaki Y, Sunaga H, Kida K (2018) Incidence of anaphylaxis associated with sugammadex. Anesth Analg. 126(5):1505–1508

    Article  CAS  PubMed  Google Scholar 

  • Money KL, Lee YJ, Elliott A (2019) Sugammadex compared with neostigmine/glycopyrrolate: an analysis of total PACU time, responsiveness, and potential for economic impact. Innov Pharm. 10:1–2

    Article  Google Scholar 

  • Murphy GS, Szokol JW, Avram MJ (2018) Neostigmine administration after spontaneous recovery to a train-of-four ratio of 0.9 to 1.0: a randomized controlled trial of the effect on neuromuscular and clinical recovery. Anaesthesiology 128:27–37

    Article  CAS  Google Scholar 

  • Naguib M, Samarkandi AH, Bakhamees HS, Magboul MA, el-Bakry AK (1995) Histamine-release haemodynamic changes produced by rocuronium, vecuronium, mivacurium, atracurium and tubocurarine. Br J Anaesth 75:588–592

    Article  CAS  PubMed  Google Scholar 

  • Oh SK, Lim BG, Park S, Yang HS, In J, Kim YB, Choi HR, Lee IO (2019) Effect of protracted dexamethasone exposure and its withdrawal on rocuronium-induced neuromuscular blockade and sugammadex reversal: an ex vivo rat study. Sci Rep 9:1–9

    Article  CAS  Google Scholar 

  • Pei DQ, Zhou HM, Zhou QH (2019) Grip strength can be used to evaluate postoperative residual neuromuscular block recovery in patients undergoing general anesthesia. Medicine (Baltimore) 98:e13940

    Article  Google Scholar 

  • Pietraszewski P, Gaszyński T (2013) Residual neuromuscular block in elderly patients after surgical procedures under general anaesthesia with rocuronium. Anaesthesiol Intensive Ther 45:77–81

    Article  PubMed  Google Scholar 

  • Raghavendra T (2002) Neuromuscular blocking drugs: discovery and development. J R Soc Med. 95:363–367

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Rahe-Meyer N, Fennema H, Schulman S, Klimscha W, Przemeck M, Blobner M (2014) Effect of reversal of neuromuscular blockade with sugammadex versus usual care on bleeding risk in a randomized study of surgical patients. Anesthesiology. 121:969–977

    Article  PubMed  Google Scholar 

  • Rodney G, Raju PK, Ball DR (2015) Not just monitoring; a strategy for managing neuromuscular blockade. Anaesthesia. 70:1105–1109

    Article  CAS  PubMed  Google Scholar 

  • Sagir O, Yucesoy Noyan F, Koroglu A, Cicek M, IlksenToprak H (2014) Comparison between the effects of rocuronium, vecuronium, and cisatracurium using train-of-four and clinical tests in elderly patients. Anesth Pain Med 2:142–148

    Article  Google Scholar 

  • Savarese JJ, Belmont MR, Hashim MA, Mook RA, Boros EE, Samano V, Patel SS, Feldman PL, Schultz JA, McNulty M, Spitzer T (2004) Preclinical pharmacology of GW280430A (AV430A) in the rhesus monkey and in the CatA comparison with mivacurium. Anesthesiology. 100(4):835–845

    Article  CAS  PubMed  Google Scholar 

  • Savarese JJ, Kitze RJ (1975) Does Clinical anesthesia Need New Neuromuscular Blocking Agents? Anesthesiology 42:236–23.

    Article  CAS  PubMed  Google Scholar 

  • Savarese JJ, McGilvra JD, Sunaga H, Belmont MR, Van Ornum SG, Savard PM (2010) Rapid chemical antagonism of neuromuscular blockade by L-cysteine adduction to and inactivation of the olefinic (double-bonded) isoquinolinium diester compounds gantacurium (AV430A), CW 002, and CW 011. Anesthesiology. 113:58–73

    Article  CAS  PubMed  Google Scholar 

  • Savarese JJ, Sunaga H, McGilvra JD, Belmont MR, Murrell MT, Jeannotte E, Cooke FE, Wastila WB, Heerdt PM (2018) Preclinical pharmacology in the rhesus monkey of CW 1759-50, a new ultra-short acting nondepolarizing neuromuscular blocking agent, degraded and antagonized by L-cysteine. Anesthesiology. 129:970–988

    Article  CAS  PubMed  Google Scholar 

  • Song IA, Seo KS, Oh AY, No HJ, Hwang JW, Jeon YT, Park SH, Do SH (2015) Timing of reversal with respect to three nerve stimulator end-points from cisatracurium-induced neuromuscular block. Anaesthesia. 70:797–802

    Article  CAS  PubMed  Google Scholar 

  • Sparr HJ, Beaufort TM, Fuchs-Buder T (2001) Drugs 61:919–921

    Article  CAS  PubMed  Google Scholar 

  • Spoerl D, Nigolian H, Czarnetzki C, Harr T (2017) Reclassifying anaphylaxis to neuromuscular blocking agents based on the presumed patho-mechanism: IgE-mediated, pharmacological adverse reaction or “Innate Hypersensitivity”? Int J Mol Sci 18:1223

    Article  PubMed Central  CAS  Google Scholar 

  • Sunaga H, Malhotra JK, Yoon E, Savarese JJ, Heerdt PM (2010b) Cysteine reversal of the novel neuromuscular blocking drug CW002 in dogs: pharmacodynamics, acute cardiovascular effects, and preliminary toxicology. Anesthesiology 112:900–913

    Article  PubMed  Google Scholar 

  • Sunaga H, Zhang Y, Savarese JJ, Emala CW (2010a) Gantacurium and CW002 do not potentiate muscarinic receptor-mediated airway smooth muscle constriction in guinea pigs. Anesthesiology 12(4):892

    Article  Google Scholar 

  • United States Food and Drug Administration. (n.d.) Notice determination that MIVACRON (Mivacurium Chloride) injection equivalent to 2 milligrams base/milliliter was not withdrawn from sale for reasons of safety or effectiveness (online). Availablefrom: https://www.federalregister.gov/documents/2007/08/08/E7-15488/determination-that-mivacron-mivacurium-chloride-injection-equivalent-to-2-milligrams-basemilliliter. (Accessed on 9 Nov 2019)

  • Vanacker BF, Vermeyen KM, Strut's MM, Rietbergen H, Vandermeersch E, Saldien V (2007) Reversal of rocuroniurn-induced neuromuscular block with the novel drug sugammadex is equally effective under maintenance anesthesia with propofol or sevoflurane. Anesth Analg. 104:563–568

    Article  CAS  PubMed  Google Scholar 

  • Zhang B, Zavalij PY, Isaacs L (2014) Acyclic CB[n]-type molecular containers: effect of solubilizing group on their function as solubilizing excipients. Org Biomol Chem. 12:2413–2422

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhang X, Cheng Q, Li L (2019) Supramolecular therapeutics to treat the side effects induced by a depolarizing neuromuscular blocking agent. Theranostics. 9:3107–3121

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are very grateful to Mrs. Shereen EL-Molla for task organization, linguistic correction, and kind help.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

SS organized the concept; wrote, reviewed, and supervised the manuscript; and designed the figures and tables. CR reviewed and shared author’s concepts and supervised the final manuscript. PA reviewed and shared author’s concepts. AE reviewed and shared author’s concepts and supervised final manuscript. All authors have read and approved the manuscript.

Corresponding author

Correspondence to Ashraf EL-Molla.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shah, S.B., Chawla, R., Pahade, A. et al. Neuromuscular blockers and their reversal: have we finally found the on-off switches?. Ain-Shams J Anesthesiol 13, 15 (2021). https://doi.org/10.1186/s42077-021-00130-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s42077-021-00130-0

Keywords